Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Oxid Med Cell Longev ; 2022: 8694462, 2022.
Article in English | MEDLINE | ID: mdl-35154573

ABSTRACT

Learning and memory disorders and decreased neuroplasticity are the main clinical manifestations of age-induced cognitive dysfunction. Orexin A (OxA) has been reported to show abnormally elevated expression in the cerebrospinal fluid (CSF) of patients with Alzheimer's disease (AD) and to be associated with cognitive impairment. Here, we further assessed whether the excitatory neurotransmitter OxA is involved in neuroplasticity and cognitive function in senescence-accelerated mouse prone 8 (SAMP8) mice. In this study, we investigated the mechanism of OxA by using behavioral tests, CSF microdialysis, immunofluorescence, toluidine blue staining, gene silencing, transmission electron microscopy, and Western blotting. The results showed that 10 Hz electroacupuncture (EA) effectively alleviated learning and memory impairment in 7-month-old SAMP8 mice, reduced OxA levels in the CSF, increased the level of the neurotransmitter glutamate, alleviated pathological damage to hippocampal tissue, improved the synaptic structure, enhanced synaptic transmission, and regulated the expression of cAMP/PKA/CREB signaling pathway-related proteins. These results suggest that EA enhances neuroplasticity in SAMP8 mice by regulating the OxA-mediated cAMP/PKA/CREB signaling pathway, thus improving cognitive function. These findings suggest that EA may be beneficial for the prevention and treatment of age-induced cognitive impairment.


Subject(s)
Aging/metabolism , Cognitive Dysfunction/therapy , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Cyclic AMP/metabolism , Electroacupuncture/methods , Neuronal Plasticity/genetics , Orexins/metabolism , Signal Transduction/genetics , Aging/genetics , Animals , Behavior, Animal , Cognition , Cognitive Dysfunction/metabolism , Disease Models, Animal , Hippocampus/metabolism , Memory Disorders/therapy , Mice , Orexins/genetics , RNA Interference , Synaptic Transmission/genetics
2.
PLoS Genet ; 16(2): e1008609, 2020 02.
Article in English | MEDLINE | ID: mdl-32097408

ABSTRACT

Neuromodulators such as monoamines are often expressed in neurons that also release at least one fast-acting neurotransmitter. The release of a combination of transmitters provides both "classical" and "modulatory" signals that could produce diverse and/or complementary effects in associated circuits. Here, we establish that the majority of Drosophila octopamine (OA) neurons are also glutamatergic and identify the individual contributions of each neurotransmitter on sex-specific behaviors. Males without OA display low levels of aggression and high levels of inter-male courtship. Males deficient for dVGLUT solely in OA-glutamate neurons (OGNs) also exhibit a reduction in aggression, but without a concurrent increase in inter-male courtship. Within OGNs, a portion of VMAT and dVGLUT puncta differ in localization suggesting spatial differences in OA signaling. Our findings establish a previously undetermined role for dVGLUT in OA neurons and suggests that glutamate uncouples aggression from OA-dependent courtship-related behavior. These results indicate that dual neurotransmission can increase the efficacy of individual neurotransmitters while maintaining unique functions within a multi-functional social behavior neuronal network.


Subject(s)
Aggression , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Neurons/metabolism , Synaptic Transmission/genetics , Vesicular Glutamate Transport Proteins/genetics , Animals , Animals, Genetically Modified , Behavior, Animal , Courtship , Drosophila Proteins/metabolism , Female , Glutamic Acid/metabolism , Male , Octopamine/metabolism , Sex Factors , Signal Transduction/genetics , Synaptic Vesicles/metabolism , Vesicular Glutamate Transport Proteins/metabolism , Vesicular Monoamine Transport Proteins/metabolism
3.
Dev Dyn ; 249(3): 328-341, 2020 03.
Article in English | MEDLINE | ID: mdl-31794137

ABSTRACT

The development of an organism from an undifferentiated single cell into a spatially complex structure requires spatial patterning of cell fates across tissues. Positional information, proposed by Lewis Wolpert in 1969, has led to the characterization of many components involved in regulating morphogen signaling activity. However, how morphogen gradients are established, maintained, and interpreted by cells still is not fully understood. Quantitative and systems-based approaches are increasingly needed to define general biological design rules that govern positional information systems in developing organisms. This short review highlights a selective set of studies that have investigated the roles of physiological signaling in modulating and mediating morphogen-based pattern formation. Similarities between neural transmission and morphogen-based pattern formation mechanisms suggest underlying shared principles of active cell-based communication. Within larger tissues, neural networks provide directed information, via physiological signaling, that supplements positional information through diffusion. Further, mounting evidence demonstrates that physiological signaling plays a role in ensuring robustness of morphogen-based signaling. We conclude by highlighting several outstanding questions regarding the role of physiological signaling in morphogen-based pattern formation. Elucidating how physiological signaling impacts positional information is critical for understanding the close coupling of developmental and cellular processes in the context of development, disease, and regeneration.


Subject(s)
Synaptic Transmission/physiology , Animals , Calcium Signaling/genetics , Calcium Signaling/physiology , Humans , Signal Transduction/genetics , Signal Transduction/physiology , Synaptic Transmission/genetics
4.
Biomed Res Int ; 2019: 2389485, 2019.
Article in English | MEDLINE | ID: mdl-31346513

ABSTRACT

Acute alcohol exposure induces unconscious condition such as coma whose main physical manifestation is the loss of righting reflex (LORR). Xingnaojing Injection (XNJI), which came from Chinese classic formula An Gong Niu Huang Pill, is widely used for consciousness disorders in China, such as coma. Although XNJI efficiently shortened the duration of LORR induced by acute ethanol, it remains unknown how XNJI acts on ethanol-induced coma (EIC). We performed experiments to examine the effects of XNJI on orexin and adenosine (AD) signaling in the lateral hypothalamic area (LHA) in EIC rats. Results showed that XNJI reduced the duration of LORR, which implied that XNJI promotes recovery form coma. Microdialysis data indicated that acute ethanol significantly increased AD release in the LHA but had no effect on orexin A levels. The qPCR results displayed a significant reduction in the Orexin-1 receptors (OX1R) expression with a concomitant increase in the A1 receptor (A1R) and equilibrative nucleoside transporter type 1 (ENT1) expression in EIC rats. In contrast, XNJI reduced the extracellular AD levels but orexin A levels remained unaffected. XNJI also counteracted the downregulation of the OX1R expression and upregulation of A1R and ENT1 expression caused by EIC. As for ADK expression, XNJI but not ethanol, displayed an upregulation in the LHA in EIC rats. Based on these results, we suggest that XNJI promotes arousal by inhibiting adenosine neurotransmission via reducing AD level and the expression of A1R and ENT1.


Subject(s)
Carrier Proteins/genetics , Coma/drug therapy , Drugs, Chinese Herbal/pharmacology , Receptor, Adenosine A1/genetics , Adenosine/genetics , Adenosine/metabolism , Animals , Coma/chemically induced , Coma/genetics , Coma/pathology , Equilibrative Nucleoside Transporter 1 , Ethanol/toxicity , Gene Expression Regulation/drug effects , Humans , Hypothalamic Area, Lateral/drug effects , Hypothalamic Area, Lateral/metabolism , Orexin Receptors/genetics , Orexins/genetics , Orexins/metabolism , Rats , Reflex, Righting/drug effects , Signal Transduction/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Wakefulness/drug effects
5.
Mol Med Rep ; 20(1): 191-197, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31115527

ABSTRACT

Dysfunctions in dopamine (DA) and serotonin (5­HT) metabolism have been widely implicated in Tourette syndrome (TS); however, the exact nature of these dysfunctions remains unclear. The objective of the present study was to investigate the variation in DA and 5­HT metabolism in a rat model of TS, and to evaluate the therapeutic effect of Ningdong granule (NDG), a traditional Chinese medicine (TCM) preparation used specifically for the treatment of TS. Rats were treated with 3,3'­iminodipropionitrile for 7 days to induce the model of TS, and were then intragastrically administered NDG each day. After 8 weeks of treatment, micro­positron emission tomography was used to measure the binding of DA D2 receptors (D2Rs), DA transporters (DATs), 5­HT2A receptors (5­HT2ARs) and 5­HT transporters (SERTs) in brain regions of interest. The results indicated that NDG could significantly reduce the typical characteristics of TS in the rat model. Decreased D2R binding and increased DAT binding were detected in the striatum compared with the binding activities in untreated rats. The density of 5­HT2AR was also significantly increased in the striatum following NDG treatment; however, SERT levels were decreased in certain brain regions, including the striatum, cortex, nucleus accumbens and amygdala. Taken together, the current results demonstrated that NDG may be effective in treating patients with TS.


Subject(s)
Dopaminergic Neurons/metabolism , Drugs, Chinese Herbal/pharmacology , Serotonergic Neurons/metabolism , Tourette Syndrome/drug therapy , Animals , Corpus Striatum/metabolism , Corpus Striatum/physiology , Disease Models, Animal , Dopamine/genetics , Dopamine Plasma Membrane Transport Proteins/genetics , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Humans , Medicine, Chinese Traditional , Nitriles/toxicity , Rats , Receptor, Serotonin, 5-HT2A/genetics , Receptors, Dopamine D2/genetics , Serotonergic Neurons/drug effects , Serotonergic Neurons/pathology , Serotonin/genetics , Serotonin Plasma Membrane Transport Proteins/genetics , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Tourette Syndrome/chemically induced , Tourette Syndrome/genetics , Tourette Syndrome/pathology
6.
Biochem Biophys Res Commun ; 509(2): 429-434, 2019 02 05.
Article in English | MEDLINE | ID: mdl-30594389

ABSTRACT

Appropriate synapse formation during development is necessary for normal brain function, and synapse impairment is often associated with brain dysfunction. Brain-derived neurotrophic factor (BDNF) and neurotrophin-3 (NT-3) are key factors in regulating synaptic development. We previously reported that BDNF/NT-3 secretion was enhanced by calcium-dependent activator protein for secretion 2 (CADPS2). Although BDNF/NT-3 and CADPS2 are co-expressed in various brain regions, the effect of Cadps2-deficiency on brain region-specific BDNF/NT-3 levels and synaptic development remains elusive. Here, we show developmental changes of BDNF/NT-3 levels and we assess disruption of excitatory/inhibitory synapses in multiple brain regions (cerebellum, hypothalamus, striatum, hippocampus, parietal cortex and prefrontal cortex) of Cadps2 knockout (KO) mice compared with wild-type (WT) mice. Compared with WT, BDNF levels in KO mice were reduced in young/adult hippocampus, but increased in young hypothalamus, while NT-3 levels were reduced in adult cerebellum and young hippocampus, but increased in adult parietal cortex. Immunofluorescence of vGluT1, an excitatory synapse marker, and vGAT, an inhibitory synapse marker, in adult KO showed that vGluT1 was higher in the cerebellum and parietal cortex but lower in the hippocampus, whereas vGAT was lower in the hippocampus and parietal cortex compared with WT. Immunolabeling for both vGluT1 and vGAT was increased in the parietal cortex but vGAT was decreased in the cerebellum in adult KO compared with WT. These data suggest that CADPS2-mediated secretion of BDNF/NT-3 may be involved in development and maturation of synapses and in the balance between inhibitory and excitatory synapses.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Calcium-Binding Proteins/genetics , Gene Expression Regulation, Developmental , Nerve Tissue Proteins/genetics , Neurons/metabolism , Neurotrophin 3/genetics , Synapses/genetics , Animals , Brain-Derived Neurotrophic Factor/metabolism , Calcium-Binding Proteins/deficiency , Cerebellum/cytology , Cerebellum/growth & development , Cerebellum/metabolism , Corpus Striatum/cytology , Corpus Striatum/growth & development , Corpus Striatum/metabolism , Hippocampus/cytology , Hippocampus/growth & development , Hippocampus/metabolism , Hypothalamus/cytology , Hypothalamus/growth & development , Hypothalamus/metabolism , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/deficiency , Neurons/cytology , Neurotrophin 3/metabolism , Organ Specificity , Parietal Lobe/cytology , Parietal Lobe/growth & development , Parietal Lobe/metabolism , Prefrontal Cortex/cytology , Prefrontal Cortex/growth & development , Prefrontal Cortex/metabolism , Synapses/classification , Synapses/metabolism , Synaptic Transmission/genetics , Vesicular Glutamate Transport Protein 1/genetics , Vesicular Glutamate Transport Protein 1/metabolism , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
7.
Sci Rep ; 8(1): 11328, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30054511

ABSTRACT

Sophorae Flavescentis Radix (SFR) is a medicinal herb with many functions that are involved in anti-inflammation, antinociception, and anticancer. SFR is also used to treat a variety of itching diseases. Matrine (MT) is one of the main constituents in SFR and also has the effect of relieving itching, but the antipruritic mechanism is still unclear. Here, we investigated the effect of MT on anti-pruritus. In acute and chronic itch models, MT significantly inhibited the scratching behavior not only in acute itching induced by histamine (His), chloroquine (CQ) and compound 48/80 with a dose-depended manner, but also in the chronic pruritus models of atopic dermatitis (AD) and acetone-ether-water (AEW) in mice. Furthermore, MT could be detected in the blood after intraperitoneal injection (i.p.) and subcutaneous injection (s.c.). Finally, electrophysiological and calcium imaging results showed that MT inhibited the excitatory synaptic transmission from dorsal root ganglion (DRG) to the dorsal horn of the spinal cord by suppressing the presynaptic N-type calcium channel. Taken together, we believe that MT is a novel drug candidate in treating pruritus diseases, especially for histamine-independent and chronic pruritus, which might be attributed to inhibition of the presynaptic N-type calcium channel.


Subject(s)
Alkaloids/administration & dosage , Antipruritics/administration & dosage , Calcium Channel Blockers/administration & dosage , Pruritus/drug therapy , Quinolizines/administration & dosage , Alkaloids/chemistry , Animals , Antipruritics/chemistry , Calcium/metabolism , Calcium Channel Blockers/chemistry , Calcium Channels, N-Type/drug effects , Calcium Channels, N-Type/genetics , Disease Models, Animal , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Male , Mice , Pruritus/genetics , Pruritus/pathology , Quinolizines/chemistry , Sophora/chemistry , Spinal Cord Dorsal Horn/drug effects , Spinal Cord Dorsal Horn/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Matrines
8.
Nat Commun ; 9(1): 504, 2018 02 05.
Article in English | MEDLINE | ID: mdl-29402935

ABSTRACT

Despite decades of research, there is a persistent debate regarding the localization of GABA/glycine neurons responsible for hyperpolarizing somatic motoneurons during paradoxical (or REM) sleep (PS), resulting in the loss of muscle tone during this sleep state. Combining complementary neuroanatomical approaches in rats, we first show that these inhibitory neurons are localized within the ventromedial medulla (vmM) rather than within the spinal cord. We then demonstrate their functional role in PS expression through local injections of adeno-associated virus carrying specific short-hairpin RNA in order to chronically impair inhibitory neurotransmission from vmM. After such selective genetic inactivation, rats display PS without atonia associated with abnormal and violent motor activity, concomitant with a small reduction of daily PS quantity. These symptoms closely mimic human REM sleep behavior disorder (RBD), a prodromal parasomnia of synucleinopathies. Our findings demonstrate the crucial role of GABA/glycine inhibitory vmM neurons in muscle atonia during PS and highlight a candidate brain region that can be susceptible to α-synuclein-dependent degeneration in RBD patients.


Subject(s)
Medulla Oblongata/physiology , Neurons/physiology , Sleep, REM/physiology , Animals , Gene Knockdown Techniques , Glycine/metabolism , Male , Medulla Oblongata/cytology , Muscle Hypotonia/physiopathology , Polysomnography , Proto-Oncogene Proteins c-fos/metabolism , REM Sleep Behavior Disorder/physiopathology , Rats, Sprague-Dawley , Synaptic Transmission/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , gamma-Aminobutyric Acid/metabolism
9.
Biol Psychiatry ; 84(1): 55-64, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29174591

ABSTRACT

BACKGROUND: Long-term synaptic plasticity is a basic ability of the brain to dynamically adapt to external stimuli and regulate synaptic strength and ultimately network function. It is dysregulated by behavioral stress in animal models of depression and in humans with major depressive disorder. Antidepressants have been shown to restore disrupted synaptic plasticity in both animal models and humans; however, the underlying mechanism is unclear. METHODS: We examined modulation of synaptic plasticity by selective serotonin reuptake inhibitors (SSRIs) in hippocampal brain slices from wild-type rats and serotonin transporter (SERT) knockout mice. Recombinant voltage-gated calcium (Ca2+) channels in heterologous expression systems were used to determine the modulation of Ca2+ channels by SSRIs. We tested the behavioral effects of SSRIs in the chronic behavioral despair model of depression both in the presence and in the absence of SERT. RESULTS: SSRIs selectively inhibited hippocampal long-term depression. The inhibition of long-term depression by SSRIs was mediated by a direct block of voltage-activated L-type Ca2+ channels and was independent of SERT. Furthermore, SSRIs protected both wild-type and SERT knockout mice from behavioral despair induced by chronic stress. Finally, long-term depression was facilitated in animals subjected to the behavioral despair model, which was prevented by SSRI treatment. CONCLUSIONS: These results showed that antidepressants protected synaptic plasticity and neuronal circuitry from the effects of stress via a modulation of Ca2+ channels and synaptic plasticity independent of SERT. Thus, L-type Ca2+ channels might constitute an important signaling hub for stress response and for pathophysiology and treatment of depression.


Subject(s)
Antidepressive Agents/therapeutic use , Calcium Channels, L-Type/metabolism , RNA-Binding Proteins/metabolism , Stress, Psychological/drug therapy , Synaptic Transmission/drug effects , Age Factors , Animals , CHO Cells , Cadmium Chloride/pharmacology , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/genetics , Cricetulus , Disease Models, Animal , Electric Stimulation , Female , Fluvoxamine/therapeutic use , HEK293 Cells , Hindlimb Suspension/psychology , Hippocampus/cytology , Humans , In Vitro Techniques , Male , Membrane Potentials/drug effects , Membrane Potentials/genetics , Nifedipine/pharmacology , Paroxetine/pharmacology , Patch-Clamp Techniques , Piperazines/pharmacology , Pyridines/pharmacology , RNA-Binding Proteins/genetics , Rats , Rats, Transgenic , Rats, Wistar , Serotonin/pharmacology , Serotonin Antagonists/pharmacology , Selective Serotonin Reuptake Inhibitors/therapeutic use , Stress, Psychological/genetics , Swimming/psychology , Synaptic Transmission/genetics , Transfection
10.
Gene ; 641: 25-34, 2018 Jan 30.
Article in English | MEDLINE | ID: mdl-29032150

ABSTRACT

Schizophrenia (SCZ) is a chronic debilitating neuropsychiatric disorder with multiple risk factors involving numerous complex genetic influences. We examined and updated a master list of clinically relevant and susceptibility genes associated with SCZ reported in the literature and genomic databases dedicated to gene discovery for characterization of SCZ genes. We used the commercially available GeneAnalytics computer-based gene analysis program and integrated genomic databases to create a molecular profile of the updated list of 608 SCZ genes to model their impact in select categories (tissues and cells, diseases, pathways, biological processes, molecular functions, phenotypes and compounds) using specialized GeneAnalytics algorithms. Genes for schizophrenia were predominantly expressed in the cerebellum, cerebral cortex, medulla oblongata, thalamus and hypothalamus. Psychiatric/behavioral disorders incorporating SCZ genes included ADHD, bipolar disorder, autism spectrum disorder and alcohol dependence as well as cancer, Alzheimer's and Parkinson's disease, sleep disturbances and inflammation. Function based analysis of major biological pathways and mechanisms associated with SCZ genes identified glutaminergic receptors (e.g., GRIA1, GRIN2, GRIK4, GRM5), serotonergic receptors (e.g., HTR2A, HTR2C), GABAergic receptors (e.g., GABRA1, GABRB2), dopaminergic receptors (e.g., DRD1, DRD2), calcium-related channels (e.g., CACNA1H, CACNA1B), solute transporters (e.g., SLC1A1, SLC6A2) and for neurodevelopment (e.g., ADCY1, MEF2C, NOTCH2, SHANK3). Biological mechanisms involving synaptic transmission, regulation of membrane potential and transmembrane ion transport were identified as leading molecular functions associated with SCZ genes. Our approach to interrogate SCZ genes and their interactions at various levels has increased our knowledge and insight into the disease process possibly opening new avenues for therapeutic intervention.


Subject(s)
Genome-Wide Association Study , Ion Transport/genetics , Membrane Potentials/genetics , Schizophrenia/genetics , Synaptic Transmission/genetics , Amino Acid Transport Systems/genetics , Calcium Channels/genetics , Cerebellum/cytology , Cerebral Cortex/cytology , Databases, Genetic , Humans , Hypothalamus/cytology , Medulla Oblongata/cytology , Receptors, Dopamine/genetics , Receptors, GABA-A/genetics , Receptors, Ionotropic Glutamate/genetics , Receptors, Serotonin/genetics , Thalamus/cytology
11.
J Comp Neurol ; 525(18): 3809-3820, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-28608460

ABSTRACT

The amount, quality, and diurnal pattern of sleep change greatly during development. Developmental changes of sleep/wake architecture are in a close relationship to brain development. The fragmentation of wake episodes is one of the salient features in the neonatal period, which is also observed in mature animals and human individuals lacking neuropeptide orexin/hypocretin signaling. This raises the possibility that developmental changes of lateral hypothalamic orexin neurons are relevant to the development of sleep/wake architecture. However, little information is available on morphological and physiological features of developing orexin neurons. To address the cellular basis for maturation of the sleep/wake regulatory system, we investigated the functional development of orexin neurons in the lateral hypothalamus. The anatomical development as well as the changes in the electrophysiological characteristics of orexin neurons was examined from embryonic to postnatal stages in orexin-EGFP mice. Prepro-orexin promoter activity was detectable at embryonic day (E) 12.0, followed by expression of orexin A after E14.0. The number of orexin neurons and their membrane capacitance reached similar levels to adults by postnatal day (P) 7, while their membrane potentials, firing rates, and action potential waveforms were developed by P21. The hyperpolarizing effect of serotonin, which is a major inhibitory signal for adult orexin neurons, was detected after E18.0 and matured at P1. These results suggest that the expression of orexin peptides precedes the maturation of electrophysiological activity of orexin neurons. The function of orexin neurons gradually matures by 3 weeks after birth, coinciding with maturation of sleep/wake architecture.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Hypothalamus , Neurons/physiology , Orexins/metabolism , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Electric Stimulation , Embryo, Mammalian , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hypothalamus/cytology , Hypothalamus/embryology , Hypothalamus/growth & development , In Vitro Techniques , Membrane Potentials/genetics , Mice , Mice, Transgenic , Neurons/metabolism , Orexins/genetics , Patch-Clamp Techniques , Serotonin/pharmacology , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , Synaptic Transmission/physiology
12.
Proc Natl Acad Sci U S A ; 114(7): 1684-1689, 2017 02 14.
Article in English | MEDLINE | ID: mdl-28143929

ABSTRACT

Experience-driven synaptic plasticity is believed to underlie adaptive behavior by rearranging the way neuronal circuits process information. We have previously discovered that O-GlcNAc transferase (OGT), an enzyme that modifies protein function by attaching ß-N-acetylglucosamine (GlcNAc) to serine and threonine residues of intracellular proteins (O-GlcNAc), regulates food intake by modulating excitatory synaptic function in neurons in the hypothalamus. However, how OGT regulates excitatory synapse function is largely unknown. Here we demonstrate that OGT is enriched in the postsynaptic density of excitatory synapses. In the postsynaptic density, O-GlcNAcylation on multiple proteins increased upon neuronal stimulation. Knockout of the OGT gene decreased the synaptic expression of the AMPA receptor GluA2 and GluA3 subunits, but not the GluA1 subunit. The number of opposed excitatory presynaptic terminals was sharply reduced upon postsynaptic knockout of OGT. There were also fewer and less mature dendritic spines on OGT knockout neurons. These data identify OGT as a molecular mechanism that regulates synapse maturity.


Subject(s)
Hypothalamus/metabolism , N-Acetylglucosaminyltransferases/metabolism , Neurons/metabolism , Synapses/metabolism , Animals , Cells, Cultured , Dendritic Spines/metabolism , Excitatory Postsynaptic Potentials/genetics , Hypothalamus/cytology , Mice, Knockout , N-Acetylglucosaminyltransferases/genetics , Neuronal Plasticity/genetics , Presynaptic Terminals/metabolism , Rats , Receptors, AMPA/genetics , Receptors, AMPA/metabolism , Synapses/genetics , Synaptic Transmission/genetics
13.
Am J Hum Genet ; 99(3): 647-665, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27499521

ABSTRACT

Homozygous loss of SMN1 causes spinal muscular atrophy (SMA), the most common and devastating childhood genetic motor-neuron disease. The copy gene SMN2 produces only ∼10% functional SMN protein, insufficient to counteract development of SMA. In contrast, the human genetic modifier plastin 3 (PLS3), an actin-binding and -bundling protein, fully protects against SMA in SMN1-deleted individuals carrying 3-4 SMN2 copies. Here, we demonstrate that the combinatorial effect of suboptimal SMN antisense oligonucleotide treatment and PLS3 overexpression-a situation resembling the human condition in asymptomatic SMN1-deleted individuals-rescues survival (from 14 to >250 days) and motoric abilities in a severe SMA mouse model. Because PLS3 knockout in yeast impairs endocytosis, we hypothesized that disturbed endocytosis might be a key cellular mechanism underlying impaired neurotransmission and neuromuscular junction maintenance in SMA. Indeed, SMN deficit dramatically reduced endocytosis, which was restored to normal levels by PLS3 overexpression. Upon low-frequency electro-stimulation, endocytotic FM1-43 (SynaptoGreen) uptake in the presynaptic terminal of neuromuscular junctions was restored to control levels in SMA-PLS3 mice. Moreover, proteomics and biochemical analysis revealed CORO1C, another F-actin binding protein, whose direct binding to PLS3 is dependent on calcium. Similar to PLS3 overexpression, CORO1C overexpression restored fluid-phase endocytosis in SMN-knockdown cells by elevating F-actin amounts and rescued the axonal truncation and branching phenotype in Smn-depleted zebrafish. Our findings emphasize the power of genetic modifiers to unravel the cellular pathomechanisms underlying SMA and the power of combinatorial therapy based on splice correction of SMN2 and endocytosis improvement to efficiently treat SMA.


Subject(s)
Endocytosis/genetics , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Actins/metabolism , Animals , Axons/pathology , Calcium/metabolism , Carrier Proteins , Disease Models, Animal , Humans , Male , Mice , Neuromuscular Junction/metabolism , Neuromuscular Junction/pathology , Oligonucleotides, Antisense , Phenotype , Presynaptic Terminals/metabolism , Pyridinium Compounds/metabolism , Quaternary Ammonium Compounds/metabolism , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/genetics , Synaptic Transmission/genetics , Zebrafish/genetics , Zebrafish/metabolism
14.
Neurobiol Aging ; 41: 73-85, 2016 May.
Article in English | MEDLINE | ID: mdl-27103520

ABSTRACT

Aging has been related to diminished cognitive function, which could be a result of ineffective synaptic function. We have previously shown that synaptic plasma membrane proteins supporting synaptic integrity and neurotransmission were downregulated in docosahexaenoic acid (DHA)-deprived brains, suggesting an important role of DHA in synaptic function. In this study, we demonstrate aging-induced synaptic proteome changes and DHA-dependent mitigation of such changes using mass spectrometry-based protein quantitation combined with western blot or messenger RNA analysis. We found significant reduction of 15 synaptic plasma membrane proteins in aging brains including fodrin-α, synaptopodin, postsynaptic density protein 95, synaptic vesicle glycoprotein 2B, synaptosomal-associated protein 25, synaptosomal-associated protein-α, N-methyl-D-aspartate receptor subunit epsilon-2 precursor, AMPA2, AP2, VGluT1, munc18-1, dynamin-1, vesicle-associated membrane protein 2, rab3A, and EAAT1, most of which are involved in synaptic transmission. Notably, the first 9 proteins were further reduced when brain DHA was depleted by diet, indicating that DHA plays an important role in sustaining these synaptic proteins downregulated during aging. Reduction of 2 of these proteins was reversed by raising the brain DHA level by supplementing aged animals with an omega-3 fatty acid sufficient diet for 2 months. The recognition memory compromised in DHA-depleted animals was also improved. Our results suggest a potential role of DHA in alleviating aging-associated cognitive decline by offsetting the loss of neurotransmission-regulating synaptic proteins involved in synaptic function.


Subject(s)
Aging/genetics , Aging/metabolism , Brain/metabolism , Docosahexaenoic Acids/metabolism , Docosahexaenoic Acids/physiology , Membrane Proteins/genetics , Membrane Proteins/metabolism , Proteome , Synapses/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Aging/psychology , Animals , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology , Down-Regulation , Fatty Acids, Omega-3/administration & dosage , Female , Male , Membrane Proteins/physiology , Memory , Mice, Inbred C57BL , Pregnancy , Recognition, Psychology
15.
J Endod ; 42(5): 752-9, 2016 May.
Article in English | MEDLINE | ID: mdl-27020968

ABSTRACT

INTRODUCTION: This study determined the gene expression profiles of the human coronal pulp (CP) and apical pulp complex (APC) with the aim of explaining differences in their functions. METHODS: Total RNA was isolated from the CP and APC, and gene expression was analyzed using complementary DNA microarray technology. Gene ontology analysis was used to classify the biological function. Quantitative reverse-transcription polymerase chain reaction and immunohistochemical staining were performed to verify microarray data. RESULTS: In the microarray analyses, expression increases of at least 2-fold were present in 125 genes in the APC and 139 genes in the CP out of a total of 33,297 genes. Gene ontology class processes found more genes related to immune responses, cell growth and maintenance, and cell adhesion in the APC, whereas transport and neurogenesis genes predominated in the CP. Quantitative reverse-transcription polymerase chain reaction and immunohistochemical staining confirmed the microarray results, with DMP1, CALB1, and GABRB1 strongly expressed in the CP, whereas SMOC2, SHH, BARX1, CX3CR1, SPP1, COL XII, and LAMC2 were strongly expressed in the APC. CONCLUSIONS: The expression levels of genes related to dentin mineralization, neurogenesis, and neurotransmission are higher in the CP in human immature teeth, whereas those of immune-related and tooth development-related genes are higher in the APC.


Subject(s)
Dental Pulp/growth & development , Gene Expression , Odontogenesis/genetics , Tooth Apex/growth & development , Adolescent , CX3C Chemokine Receptor 1 , Calbindin 1/genetics , Calcium-Binding Proteins/genetics , Cell Adhesion/genetics , Child , Child, Preschool , Collagen Type XII/genetics , Dental Pulp/anatomy & histology , Dental Pulp/cytology , Dental Pulp/diagnostic imaging , Extracellular Matrix Proteins/genetics , Female , Gene Expression Profiling , Hedgehog Proteins/genetics , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , Laminin/genetics , Male , Microarray Analysis/methods , Neurogenesis/genetics , Osteopontin/genetics , Phosphoproteins/genetics , RNA/analysis , Real-Time Polymerase Chain Reaction/methods , Receptors, Chemokine/genetics , Receptors, GABA-A/genetics , Republic of Korea , Synaptic Transmission/genetics , Tooth Apex/anatomy & histology , Tooth Apex/cytology , Tooth Apex/diagnostic imaging , Tooth Calcification/genetics , Transcription Factors/genetics , Young Adult
16.
Proc Natl Acad Sci U S A ; 113(10): E1382-91, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26903620

ABSTRACT

Higher-level cognitive processes strongly depend on a complex interplay between mediodorsal thalamus nuclei and the prefrontal cortex (PFC). Alteration of thalamofrontal connectivity has been involved in cognitive deficits of schizophrenia. Prefrontal serotonin (5-HT)2A receptors play an essential role in cortical network activity, but the mechanism underlying their modulation of glutamatergic transmission and plasticity at thalamocortical synapses remains largely unexplored. Here, we show that 5-HT2A receptor activation enhances NMDA transmission and gates the induction of temporal-dependent plasticity mediated by NMDA receptors at thalamocortical synapses in acute PFC slices. Expressing 5-HT2A receptors in the mediodorsal thalamus (presynaptic site) of 5-HT2A receptor-deficient mice, but not in the PFC (postsynaptic site), using a viral gene-delivery approach, rescued the otherwise absent potentiation of NMDA transmission, induction of temporal plasticity, and deficit in associative memory. These results provide, to our knowledge, the first physiological evidence of a role of presynaptic 5-HT2A receptors located at thalamocortical synapses in the control of thalamofrontal connectivity and the associated cognitive functions.


Subject(s)
Association Learning/physiology , Cerebral Cortex/physiology , Neuronal Plasticity/physiology , Receptor, Serotonin, 5-HT2A/physiology , Thalamus/physiology , Animals , Blotting, Western , Cerebral Cortex/metabolism , Electrophysiological Phenomena , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence , Neuronal Plasticity/genetics , Prefrontal Cortex/metabolism , Prefrontal Cortex/physiology , Protein Kinase C/metabolism , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2A/genetics , Receptor, Serotonin, 5-HT2A/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/metabolism , Synapses/physiology , Synaptic Transmission/genetics , Synaptic Transmission/physiology , Thalamus/metabolism , Type C Phospholipases/metabolism
17.
Neuron ; 88(5): 879-891, 2015 Dec 02.
Article in English | MEDLINE | ID: mdl-26606997

ABSTRACT

Exogenously expressed opsins are valuable tools for optogenetic control of neurons in circuits. A deeper understanding of neural function can be gained by bringing control to endogenous neurotransmitter receptors that mediate synaptic transmission. Here we introduce a comprehensive optogenetic toolkit for controlling GABA(A) receptor-mediated inhibition in the brain. We developed a series of photoswitch ligands and the complementary genetically modified GABA(A) receptor subunits. By conjugating the two components, we generated light-sensitive versions of the entire GABA(A) receptor family. We validated these light-sensitive receptors for applications across a broad range of spatial scales, from subcellular receptor mapping to in vivo photo-control of visual responses in the cerebral cortex. Finally, we generated a knockin mouse in which the "photoswitch-ready" version of a GABA(A) receptor subunit genomically replaces its wild-type counterpart, ensuring normal receptor expression. This optogenetic pharmacology toolkit allows scalable interrogation of endogenous GABA(A) receptor function with high spatial, temporal, and biochemical precision.


Subject(s)
Brain/cytology , Neural Inhibition/physiology , Optogenetics/methods , Receptors, GABA-A/metabolism , Synaptic Transmission/physiology , Animals , Binding Sites/drug effects , Binding Sites/physiology , Cells, Cultured , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , In Vitro Techniques , Mice, Knockout , Mutation/genetics , Neural Inhibition/drug effects , Patch-Clamp Techniques , Phosphines/pharmacology , Photic Stimulation , Receptors, GABA-A/genetics , Synapsins/genetics , Synapsins/metabolism , Synaptic Transmission/drug effects , Synaptic Transmission/genetics , gamma-Aminobutyric Acid/pharmacology
18.
Endocrinology ; 156(8): 2831-42, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26061727

ABSTRACT

Corticosteroids act classically via cognate nuclear receptors to regulate gene transcription; however, increasing evidence supports rapid, nontranscriptional corticosteroid actions via activation of membrane receptors. Using whole-cell patch clamp recordings in hypothalamic slices from male mouse genetic models, we tested for nongenomic glucocorticoid actions at glutamate and gamma aminobutyric acid (GABA) synapses in hypothalamic neuroendocrine cells, and for their dependence on the nuclear glucocorticoid receptor (GR). In enhanced green fluorescent protein-expressing CRH neurons of the paraventricular nucleus (PVN) and in magnocellular neurons of the PVN and supraoptic nucleus (SON), dexamethasone activated postsynaptic membrane-associated receptors and G protein signaling to elicit a rapid suppression of excitatory postsynaptic inputs, which was blocked by genetic deletion of type I cannabinoid receptors and a type I cannabinoid receptor antagonist. In magnocellular neurons, dexamethasone also elicited a rapid nitric oxide-dependent increase in inhibitory postsynaptic inputs. These data indicate a rapid, synapse-specific glucocorticoid-induced retrograde endocannabinoid signaling at glutamate synapses and nitric oxide signaling at GABA synapses. Unexpectedly, the rapid glucocorticoid effects on both excitatory and inhibitory synaptic transmission were lost with conditional deletion of GR in the PVN and SON in slices from a single minded-1-cre-directed conditional GR knockout mouse. Thus, the nongenomic glucocorticoid actions at glutamate and GABA synapses on PVN and SON neuroendocrine cells are dependent on the nuclear GR. The nuclear GR, therefore, is responsible for transducing the rapid steroid response at the membrane, or is either a critical component in the signaling cascade or regulates a critical component of the signaling cascade of a distinct membrane GR.


Subject(s)
Glucocorticoids/pharmacology , Hypothalamus/drug effects , Neuroendocrine Cells/drug effects , Receptors, Glucocorticoid/physiology , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuroendocrine Cells/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Receptors, Glucocorticoid/genetics , Supraoptic Nucleus/metabolism , Synaptic Transmission/genetics , Time Factors
19.
Mol Psychiatry ; 20(4): 509-19, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25092243

ABSTRACT

Compelling evidence supports the important role of the glutamatergic system in the pathophysiology of major depression and also as a target for rapid-acting antidepressants. However, the functional role of glutamate release/transmission in behavioral processes related to depression and antidepressant efficacy remains to be elucidated. In this study, glutamate release and behavioral responses to tail suspension, a procedure commonly used for inducing behavioral despair, were simultaneously monitored in real time. The onset of tail suspension stress evoked a rapid increase in glutamate release in hippocampal field CA3, which declined gradually after its offset. Blockade of N-methyl-D-aspartic acid (NMDA) receptors by intra-CA3 infusion of MK-801, a non-competitive NMDA receptor antagonist, reversed behavioral despair. A subpopulation of granule neurons that innervated the CA3 region expressed leptin receptors and these cells were not activated by stress. Leptin treatment dampened tail suspension-evoked glutamate release in CA3. On the other hand, intra-CA3 infusion of NMDA blocked the antidepressant-like effect of leptin in reversing behavioral despair in both the tail suspension and forced swim tests, which involved activation of Akt signaling in DG. Taken together, these results suggest that the DG-CA3 glutamatergic pathway is critical for mediating behavioral despair and antidepressant-like responses to leptin.


Subject(s)
CA3 Region, Hippocampal/pathology , Dentate Gyrus/pathology , Depression/drug therapy , Glutamic Acid/metabolism , Leptin/therapeutic use , N-Methylaspartate/metabolism , Synaptic Transmission/drug effects , Animals , Antidepressive Agents/therapeutic use , CA3 Region, Hippocampal/drug effects , Dentate Gyrus/drug effects , Depression/pathology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Agents/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , N-Methylaspartate/pharmacology , RNA, Untranslated/genetics , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Synapses/drug effects , Synaptic Transmission/genetics , Wakefulness
20.
Science ; 344(6188): 1178-82, 2014 Jun 06.
Article in English | MEDLINE | ID: mdl-24904170

ABSTRACT

Auditory hallucinations in schizophrenia are alleviated by antipsychotic agents that inhibit D2 dopamine receptors (Drd2s). The defective neural circuits and mechanisms of their sensitivity to antipsychotics are unknown. We identified a specific disruption of synaptic transmission at thalamocortical glutamatergic projections in the auditory cortex in murine models of schizophrenia-associated 22q11 deletion syndrome (22q11DS). This deficit is caused by an aberrant elevation of Drd2 in the thalamus, which renders 22q11DS thalamocortical projections sensitive to antipsychotics and causes a deficient acoustic startle response similar to that observed in schizophrenic patients. Haploinsufficiency of the microRNA-processing gene Dgcr8 is responsible for the Drd2 elevation and hypersensitivity of auditory thalamocortical projections to antipsychotics. This suggests that Dgcr8-microRNA-Drd2-dependent thalamocortical disruption is a pathogenic event underlying schizophrenia-associated psychosis.


Subject(s)
22q11 Deletion Syndrome/genetics , Auditory Cortex/metabolism , Haploinsufficiency , RNA-Binding Proteins/genetics , Receptors, Dopamine D2/biosynthesis , Schizophrenia/genetics , Thalamus/metabolism , 22q11 Deletion Syndrome/drug therapy , Animals , Antipsychotic Agents/therapeutic use , Disease Models, Animal , Drug Resistance/genetics , Mice , Mice, Mutant Strains , MicroRNAs/metabolism , Receptors, Dopamine D2/genetics , Schizophrenia/drug therapy , Synaptic Transmission/genetics
SELECTION OF CITATIONS
SEARCH DETAIL